PP2A Activators Enhance Efficacy of FLT3 Inhibitors in FLT3-ITD Acute Myeloid Leukemia Cells through AKT Inactivation-Dependent Pim-1 and c-Myc Proteasomal Degradation

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1276-1276
Author(s):  
Mario Scarpa ◽  
Prerna Singh ◽  
Shivani Kapoor ◽  
Jonelle K. Lee ◽  
Sandrine Niyongere ◽  
...  

Introduction fms-like tyrosine like kinase 3 internal tandem duplication (FLT3-ITD), present in acute myeloid leukemia (AML) cells of 30% of patients, results in constitutive and aberrant FLT3 signaling and, clinically, short disease-free survival. Efficacy of FLT3 inhibitors is limited and transient, but may be enhanced by dual targeting of FLT3-ITD signaling pathways. The tumor suppressor protein phosphatase 2A (PP2A) is inhibited in cells with FLT3-ITD. The oncogenic serine/threonine kinase Pim-1 is transcriptionally upregulated and also stabilized by PP2A inhibition in cells with FLT3-ITD. Pim-1 contributes directly to FLT3-ITD proliferative and anti-apoptotic effects, and also phosphorylates and stabilizes FLT3-ITD in a positive feedback loop. Moreover FLT3-ITD, PP2A and Pim-1 all regulate the transcription factor c-Myc. PP2A-activating drugs enhance efficacy of FLT3 inhibitors. We sought to identify mechanisms underlying the efficacy of this combination. Methods Ba/F3-ITD and MV4-11 cells, with FLT3-ITD, and blasts from patients with AML with FLT3-ITD were cultured with the FLT3 inhibitors gilteritinib (15 nM) or quizartinib (1 nM) and/or the PP2A-activating drugs FTY720 (2-4 µM) or DT-061 (10 µM), or with DMSO control. Pim-1, c-Myc, p-AKT (S473 and T308) and AKT protein expression was measured by immunoblotting, along with p-STAT5 (Y694), STAT5, p-PP2A (Y307) and PP2A expression. To study post-translational regulation, cells were cultured with cycloheximide (100 µg/mL) with and without the proteasome inhibitor MG-132 (20 µM). Ubiquitinated c-Myc was measured by co-immunoprecipitation and immunoblotting with c-Myc and ubiquitin antibodies. Ba/F3-ITD cells were stably transfected with estrogen receptor (ER)-c-Myc, kinase-dead Pim-1 or myristoylated AKT plasmids or corresponding empty vectors. Apoptosis was detected by Annexin V and propidium iodine staining, measured by flow cytometry. Cells were also cultured with the pan-Pim kinase inhibitor AZD1208 (1 µM), the Myc inhibitor 10058-F4 (100 µM) or the pan-AKT inhibitor MK-2206 (5 µM). Results Concurrent treatment of Ba/F3-ITD and MV4-11 cells and primary AML cells with FLT3-ITD with a FLT3 inhibitor (gilteritinib or quizartinib) and a PP2A-activating drug (FTY720 or DT-061) decreased growth and increased apoptosis induction, relative to treatment with single drugs. Concurrent FLT3 inhibitor and PP2A-activating drug treatment decreased expression of both Pim-1 and c-Myc protein. Concurrent treatment decreased Pim-1 half-life from 15 to 5 minutes, and c-Myc half-life from 30 to 5 minutes, while half-lives were restored by concurrent treatment with the proteasome inhibitor MG-132. Concurrent treatment was also shown to increase c-Myc ubiquitination. Effects of concurrent treatment on Pim-1 and c-Myc were independent, as transfection with kinase-dead Pim-1 or treatment with Pim inhibitor AZD1208 did not alter c-Myc downregulation, and c-Myc overexpression or treatment with Myc inhibitor 10058-F4 did not alter Pim-1 downregulation. Concurrent treatment with FLT3 inhibitor and PP2A-activating drug did not alter expression of c-Myc deubiquitinases, but rapidly decreased AKT S473 and T308 phosphorylation. FLT3 inhibitor and PP2A activator co-treatment did not induce downregulation or increased turnover of Pim-1 and c-Myc protein or apoptosis in cells with constitutive AKT activation caused by transfection of myristoylated AKT. Moreover, AKT inhibition downregulated Pim-1 and c-Myc protein expression, decreased Pim-1 and c-Myc protein half-lives from 15 to 5 minutes and 30 to 10 minutes, respectively, and induced apoptosis of cells with FLT3-ITD, replicating the effects of FLT3 inhibitor and PP2A activator co-treatment. Conclusion PP2A activators enhance the efficacy of FLT3 inhibitors in AML cells with FLT3-ITD through AKT inactivation-dependent increased Pim-1 and c-Myc proteasomal degradation, which is a novel mechanism. The data support further preclinical and clinical testing of this dual targeting approach to treatment of AML with FLT3-ITD. Disclosures Baer: Takeda: Research Funding; Incyte: Research Funding; Kite: Research Funding; Forma: Research Funding; AI Therapeutics: Research Funding; Abbvie: Research Funding; Astellas: Research Funding.

2016 ◽  
Vol 113 (43) ◽  
pp. E6669-E6678 ◽  
Author(s):  
Mark A. Gregory ◽  
Angelo D’Alessandro ◽  
Francesca Alvarez-Calderon ◽  
Jihye Kim ◽  
Travis Nemkov ◽  
...  

Activating mutations in FMS-like tyrosine kinase 3 (FLT3) are common in acute myeloid leukemia (AML) and drive leukemic cell growth and survival. Although FLT3 inhibitors have shown considerable promise for the treatment of AML, they ultimately fail to achieve long-term remissions as monotherapy. To identify genetic targets that can sensitize AML cells to killing by FLT3 inhibitors, we performed a genome-wide RNA interference (RNAi)-based screen that identified ATM (ataxia telangiectasia mutated) as being synthetic lethal with FLT3 inhibitor therapy. We found that inactivating ATM or its downstream effector glucose 6-phosphate dehydrogenase (G6PD) sensitizes AML cells to FLT3 inhibitor induced apoptosis. Examination of the cellular metabolome showed that FLT3 inhibition by itself causes profound alterations in central carbon metabolism, resulting in impaired production of the antioxidant factor glutathione, which was further impaired by ATM or G6PD inactivation. Moreover, FLT3 inhibition elicited severe mitochondrial oxidative stress that is causative in apoptosis and is exacerbated by ATM/G6PD inhibition. The use of an agent that intensifies mitochondrial oxidative stress in combination with a FLT3 inhibitor augmented elimination of AML cells in vitro and in vivo, revealing a therapeutic strategy for the improved treatment of FLT3 mutated AML.


2019 ◽  
Vol 51 (5) ◽  
pp. 478-483
Author(s):  
Cade D Arries ◽  
Sophia L Yohe

Abstract Background FMS-like tyrosine kinase-3 (FLT3-ITD) mutations are some of the most common mutations in acute myeloid leukemia (AML), and patient outcomes have improved since the advent of tyrosine kinase inhibitors. First, granulocytic differentiation was described in FLT3-positive AML treated with FLT3 inhibitors, and more recently, monocytic differentiation was reported. Methods Two patients with myelomonocytic cells in their bone marrow were identified during routine follow-up after AML treatment that included FLT3 inhibitors. The bone marrow study was done as standard of care. Results Both patients had FLT3-ITD+ AML and showed an atypical maturing monocytic cell population and a decrease in the leukemic blast cell population after FLT3 inhibitor therapy. Concurrent genetic testing revealed persistent genetic abnormalities. Conclusions These cases illustrate monocytic maturation in FLT3+ AML after FLT3 inhibitor treatment. It is critical for pathologists and clinicians to be aware of the differentiation phenomenon, as these patients have persistent molecular abnormalities despite response to treatment and normalization of blast counts.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3949-3949
Author(s):  
Paolo Strati ◽  
Hagop M Kantarjian ◽  
Aziz Nazha ◽  
Gautam Borthakur ◽  
Naval G. Daver ◽  
...  

Abstract Background Acute Myeloid Leukemia (AML) and Myelodysplastic Syndrome (MDS) affect primarily elderly pts. Their treatment with aggressive chemotherapy is frequently challenging. Moreover, pts with FLT3 mutations have very poor prognosis. We hypothesized that the combination of midostaurin, a FLT3 inhibitor, and 5-AZA, a hypomethylating agent, may be an effective and safe regimen. Methods Both untreated (8) and previously treated (36) pts with AML or MDS were eligible for this study, regardless of FLT3 mutation and prior exposure to FLT3 inhibitors. Pts received 5-AZA 75 mg/mq subcutaneously or intravenously on day 1-7 and midostaurin 25 mg bid (in cohort 1 of phase I) or 50 mg bid (in cohort 2 of Phase I and in Phase II) orally on day 8-21 during the first cycle and continuously thereafter, for 12 cycles of 28 days duration. Cytogenetic risk was defined according to MRC criteria. Differences between categorical variables were compared by the chi2 test. CR duration (CRD) was calculated from the time of CR achievement until relapse and estimated by the Kaplan-Meier method and compared by the log-rank test. Results Fourty-four pts were enrolled, 13 included in Phase I and 31 in Phase II. Baseline pts’ characteristics are shown in the Table. Thirty-eight pts (86%) received 50 mg bid of midostaurin, and 6 (14%; Phase I) received 25 mg bid. The median number of administered cycles was 2 (1-9). Grade 3-4 hematological toxicities consisted of 95% neutropenia, 64% anemia and 93% thrombocytopenia. Grade 3-4 non-hematological toxicities consisted of 45% infections, 23% hypokalemia, 16% hyponatremia, 7% reduction in ejection fraction, 7% hyperuricemia, 4% hyperglycemia, 4% nausea/vomiting, 4% QTc prolongation, 4% hyperbilirubinemia, and 4% elevated AST. Eleven pts (25%) achieved a CR, 9 with incomplete platelet recovery (20%), after a median time of 13 (10-16) weeks from treatment start. Five (11%) of these pts relapsed after achieving CR. Two pts (5%) received an allogeneic stem cell transplant while on study, one in CR and one primary refractory (after a blast count drop from 27 to 7%), and they are both still in CR and alive. Among 26 pts with FLT3 ITD and no D835 mutation, 9 (35%) achieved CR/CRp. Six of 18 (33%) pts not previously exposed to FLT3 inhibitors responded. There was no significant correlation of dose with response (24% with 50 mg bid vs 33% with 25 mg bid, p=0.63). After a median follow-up of 15 (3-72) weeks, 20 pts (64%) died, 3 (7%) while on study (2 died of sepsis, 1 of unknown causes with progressive disease). The median CRD was 16 (9-23) months. Factors significantly associated with a longer CRD were male sex (p=0.04), age older than 65 years (0.03) and use of 50 mg bid of midostaurin (p=0.02). Conclusions The combination of midostaurin and 5-AZA is safe and well tolerated. Its efficacy is most noticeable among pts with FLT3 mutations. A longer response duration is observed using midostaurin at 50 mg bid dose and in elderly male pts. Disclosures: Ravandi: CELGENE: Honoraria; NOVARTIS: Honoraria. Cortes:ARIAD: Consultancy, Research Funding; ASTELLAS: Research Funding; AMBIT: Research Funding; AROG: Research Funding; NOVARTIS: Research Funding.


Blood ◽  
2011 ◽  
Vol 117 (12) ◽  
pp. 3286-3293 ◽  
Author(s):  
Takashi Sato ◽  
Xiaochuan Yang ◽  
Steven Knapper ◽  
Paul White ◽  
B. Douglas Smith ◽  
...  

AbstractWe examined in vivo FLT3 inhibition in acute myeloid leukemia patients treated with chemotherapy followed by the FLT3 inhibitor lestaurtinib, comparing newly diagnosed acute myeloid leukemia patients with relapsed patients. Because we noted that in vivo FLT3 inhibition by lestaurtinib was less effective in the relapsed patients compared with the newly diagnosed patients, we investigated whether plasma FLT3 ligand (FL) levels could influence the efficacy of FLT3 inhibition in these patients. After intensive chemotherapy, FL levels rose to a mean of 488 pg/mL on day 15 of induction therapy for newly diagnosed patients, whereas they rose to a mean of 1148 pg/mL in the relapsed patients. FL levels rose even higher with successive courses of chemotherapy, to a mean of 3251 pg/mL after the fourth course. In vitro, exogenous FL at concentrations similar to those observed in patients mitigated FLT3 inhibition and cytotoxicity for each of 5 different FLT3 inhibitors (lestaurtinib, midostaurin, sorafenib, KW-2449, and AC220). The dramatic increase in FL level after chemotherapy represents a possible obstacle to inhibiting FLT3 in this clinical setting. These findings could have important implications regarding the design and outcome of trials of FLT3 inhibitors and furthermore suggest a rationale for targeting FL as a therapeutic strategy.


2021 ◽  
Author(s):  
Dan Xu ◽  
Zhao Yin ◽  
Ying Yang ◽  
Yishan Chen ◽  
Changfen Huang ◽  
...  

Abstract Background: Autophagy plays a critical role in drug resistance in acute myeloid leukemia (AML), including the subtype with FLT3-ITD mutation. Yet how autophagy is activated and mediates resistance to FLT3 inhibitors in FLT3-ITD-positive AML remains unsure. Methods: We detected the alteration of autophagy in FLT3-ITD-positive leukemic cells after versus before acquired resistance to FLT3 inhibitors; tested the stimulative effect of acquired D835Y mutation and bone marrow micro-environment (BME) on autophagy; explored the mechanism of autophagy mediating FLT3 inhibitor resistance. Results: Sorafenib-resistant cells markedly overexpressed autophagy in comparison with sorafenib-sensitive cells or the cells before sorafenib treatment. Both acquired D835Y mutation and BME activated cytoprotective autophagy to induce FLT3 inhibitor resistance. Autophagy activation decreased the suppression efficacy of FLT3 inhibitors on FLT3 downstream signaling and then weakened their anti-leukemia effect. Inhibition of autophagy with CQ significantly enhanced the suppressive effect of FLT3 inhibitor on FLT3 downstream signaling, in the end overcame FLT3 inhibitor resistance. Conclusions: Autophagy might be stimulated by acquired mutation or BME, and bypass activate FLT3 downstream signaling to mediate FLT3 inhibitor resistance in FLT3-ITD-positive AML. Targeting autophagy could be a promising strategy to overcome resistance.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. TPS7062-TPS7062
Author(s):  
Farhad Ravandi ◽  
Stephen Anthony Strickland ◽  
Scott R. Solomon ◽  
Aziz Nazha ◽  
Roland B. Walter ◽  
...  

TPS7062 Background: FLT3-ITD is one of the most common genetic lesions in acute myeloid leukemia (AML). PIM kinases are oncogenic FLT3-ITD targets expressed in AML cells and increased PIM kinase expression is found in relapse samples from AML patients treated with FLT3 inhibitors. In addition, inhibition of PIM kinases restores sensitivity to FLT3 inhibitors and dual FLT3/PIM inhibition eradicates FLT3-ITD+ cells including primary AML cells. SEL24/MEN1703, a potent PIM/FLT3 dual inhibitor, demonstrates a significantly broader spectrum of activity in AML cell lines and primary AML blasts, irrespective of FLT3 status, compared to monotherapy with either FLT3 or PIM inhibitors such as quizartinib or AZD1208. Methods: CLI24-001 is a First in Human, open label, non-randomized, multi-center, Phase I/II dose-escalation and cohort expansion study of SEL24/MEN1703 in AML patients (excluding APL) not suitable for chemotherapy. SEL24/MEN1703 is given orally, QD, for 14 days in a 21-day cycle with cycles repeated until disease progression or unacceptable toxicity. Dose escalation follows a 3+3 design to identify the recommended phase 2 dose (RP2D). In the phase 2 part/cohort expansion, subjects will receive SEL24/MEN1703 at the RP2D, to further investigate the safety profile and signs of antileukemic activity. In both study parts, patients are eligible regardless of mutational status and/or prior exposure to FLT3 inhibitors; prior treatment with PIM inhibitors is not allowed. Main inclusion criteria comprise a white blood count (WBC) of ≤30 x 109/L (hydroxyurea/leukoapheresis permitted to lower WBC). Key secondary objectives include pharmacokinetics (PK) and single agent efficacy. The study is enrolling at 5 US sites and will be extended, both in US and EU, in the cohort expansion part. This is the first trial testing a dual PIM/FLT3 inhibitor with the potential to be active in AML regardless of FLT3 status andwith a potential to overcome FLT3 inhibitor resistance. (Sci Adv. 2015;1:e1500221; Oncotarget. 2018 Mar 30;9(24):16917-16931) Clinical trial information: NCT03008187.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2597-2597
Author(s):  
Kensuke Kojima ◽  
Marina Konopleva ◽  
Twee Tsao ◽  
Michael Andreeff ◽  
Hiroshi Ishida ◽  
...  

Abstract Abstract 2597 Poster Board II-573 Introduction: Activating mutations of the Fms-like tyrosine kinase-3 gene (FLT3) occur in approximately 30–40% of acute myeloid leukemia (AML) patients. FLT3 mutations confer numerous oncogenic properties, including dysregulated proliferation, resistance to apoptosis and a block in differentiation. FLT3 mutations result in abnormal activation of the downstream pathways, including signal transducer and activator of transcription 5 (STAT5), mitogen-activated protein kinase kinase (Mek)/extracellular signal–regulated kinase (Erk) and phosphatidylinositol-3 kinase (PI3K)/Akt. Activation of these downstream effectors has been thought to allow leukemia cells to evade apoptosis. Targeting of FLT3 mutations is a promising approach to overcome the dismal prognosis of acute myeloid leukemia (AML) with activating FLT3 mutations. Current trials are combining FLT3 inhibitors with p53-activating conventional chemotherapy. The mechanisms of cytotoxicity of FLT3 inhibitors are poorly understood. We investigated the interaction of FLT3 and p53 pathways after their simultaneous blockade using the selective FLT3 inhibitor FI-700 and the MDM2 inhibitor Nutlin-3 in AML. Results: FI-700 induced G1-phase cell cycle arrest and apoptosis as evidenced by increased sub-G1 DNA content and phosphatidylserine externalization in FLT3/ITD MOLM-13 (FLT3-ITD, wild-type (wt)-p53) and MV4-11NR (FLT3-ITD, mutated-p53) AML cells. FI-700 did not affect cell cycle distribution patterns nor did it induce apoptosis in FLT3/WT OCI-AML-3 (FLT3/WT, wt-p53) and HL-60 (FLT3/WT, del (del)-p53). Wt-p53 MOLM-13 and OCI-AML-3 cells were susceptible to Nutlin-induced apoptosis. FI-700 augmented Nutlin-induced Bax activation, mitochondrial membrane potential (MMP) loss, caspase-3 activation and phosphatidylserine externalization in MOLM-13 cells. FI-700 rapidly reduced Mcl-1 levels in FLT3/ITD cells, mainly by enhancing proteasomal Mcl-1 degradation. Levels of other Bcl-2 family proteins examined did not change significantly. Mcl-1 levels were only modestly reduced upon Nutlin treatment. The FI-700/Nutlin-3 combination profoundly reduced Mcl-1 levels. Immunoprecipitation/ immunoblotting results suggested that the drug combination results in a profound decrease in Mcl-1-bound Bim. FI-700 enhanced doxorubicin-induced apoptosis in FLT3/ITD MOLM-13 and MV4-11NR cells, suggesting that FI-700 can enhance both the p53-dependent and the p53-independent apoptotic effects of doxorubicin. Finally, cooperative apoptotic effects of FI-700/Nutlin-3 were seen in primary AML cells with FLT3/ITD. Conclusion: FLT3 inhibition by FI-700 immediately reduces anti-apoptotic Mcl-1 levels and enhances Nutlin-induced p53-mediated mitochondrial apoptosis in FLT3/ITD-expressing AML cells via the Mcl-1/Noxa axis. FLT3 inhibition, in combination with p53-inducing agents, might represent a potential therapeutic approach in AML with FLT3/ITD. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4059-4059
Author(s):  
Philip C. Amrein ◽  
Eyal C. Attar ◽  
Traci M. Blonquist ◽  
Andrew M. Brunner ◽  
Gabriela S. Hobbs ◽  
...  

Abstract Introduction: Treatment of acute myeloid leukemia (AML) has remained largely unchanged for several decades despite the emergence of new agents. Long-term survival for patients aged >60 years is less than 10% (median survival 10.5 months). Targeting the proteasome in treating AML is attractive, since leukemia stem cells have demonstrated sensitivity to proteasome inhibition, perhaps through down regulation of nuclear NF-KB (Guzman, Blood 2001). Preclinical studies in leukemia cell lines revealed synergistic cytotoxicity when bortezomib, a proteasome inhibitor, was combined with the standard agents daunorubicin and cytarabine. We have shown that adding bortezomib to standard treatment in AML results in a high remission rate, although neurotoxicity was noted among treated patients, 12% grade 3 sensory (Attar, …, Amrein, et al. Clin Cancer Res 2008, Attar, … Amrein, J Clin Oncol 2012). The next generation proteasome inhibitor, ixazomib, which is less frequently associated with neurotoxicity, was therefore selected for combination with conventional chemotherapy in this phase I trial. The primary objective was to determine the maximum tolerated dose (MTD) in the combination, initially in induction, and then in combination with consolidation in a subsequent portion of the overall study. We report here the results of the induction portion of the study, which has been completed. Methods: Adults >60 years of age with newly diagnosed AML were screened for eligibility. Patients with secondary AML were eligible, including those with prior hypomethylating agent therapy for myelodysplastic syndromes (MDS). We excluded those with promyelocytic leukemia. The induction treatment consisted of the following: cytarabine 100 mg/m2/day by continuous IV infusion, Days 1-7; daunorubicin 60 mg/m2/day IV, Days 1, 2, 3; ixazomib orally at the cohort dose, Days 2, 5, 9, and 12 A standard 3 + 3 patient cohort dose escalation design was used to determine whether the dose of ixazomib could be safely escalated in 3 cohorts (1.5 mg/day, 2.3 mg/day, 3.0 mg/day), initially in induction and subsequently in consolidation. The dose of 3.0 mg/day was the maximum planned for this study. The determined MTD of ixazomib in the first portion of the trial would be used during induction in the second portion, which seeks to test dose escalation of ixazomib during consolidation. Secondary objectives included rate of complete remission and disease-free survival. Results: Fourteen patients have been analyzed for toxicity and activity during the induction portion of the study. There were 4 (28%) patients with either secondary AML or treatment related AML, 9 (64%) were male, and the median age was 67 years (range 62-80 years). There have been no grade 5 toxicities due to study drug. Three patients died early due to leukemia, 2 of which were replaced for assessment of the MTD. Nearly all the grade 3 and 4 toxicities were hematologic (Table). There was 1 DLT (grade 3 thrombocytopenia) indicated at the highest dose level. There has been no neurotoxicity with ixazomib to date. Among the 14 patients, there have been 10 complete remissions (CR's) and 1 CRi for a remission rate of 79%. Conclusions: The highest dose level planned for this portion of the trial, 3.0 mg of ixazomib, was reached with 1 DLT and is the recommended dose for induction in the next portion of this study, which will seek to determine a safe ixazomib dose in combination with conventional consolidation therapy. That no neurotoxicity was encountered was reassuring, and the remission rate in this older adult population is favorable. Table. Table. Disclosures Amrein: Takeda: Research Funding. Attar:Agios: Employment, Equity Ownership. Brunner:Takeda: Research Funding; Novartis: Research Funding; Celgene: Consultancy, Research Funding. Fathi:Celgene: Consultancy, Honoraria, Research Funding; Boston Biomedical: Consultancy, Honoraria; Astellas: Honoraria; Agios: Honoraria, Research Funding; Jazz: Honoraria; Seattle Genetics: Consultancy, Honoraria; Takeda: Consultancy, Honoraria.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4940-4940 ◽  
Author(s):  
Adam Cloe ◽  
Richard A. Larson ◽  
Jason X. Cheng

Abstract Introduction FLT3 is a receptor tyrosine kinase that plays a role in hematopoietic stem/progenitor cell proliferation and survival and is frequently found to be mutated in patients with acute myeloid leukemia (AML). Mutations that lead to constitutive activation of FLT3 (such as internal tandem duplications of the juxtamembrane domain or point mutations involving the kinase domain) are associated with a poor prognosis. This poor prognosis is in part due to an increased relapse rate after allogeneic hematopoietic cell transplant (HCT). Several compounds that inhibit the activity of FLT3 in vitro, including ASP2215, midostaurin and quizartinib, are now being studied in clinical trials for the treatment of AML. Inhibition of the formation of phospho-FLT3 has been correlated with clinical anti-leukemia effects and remissions. Methods In this pilot study we use immunohistochemistry to measure the levels of activated FLT3 in bone marrow biopsies of patients prior to and during treatment inclinical trials at our institution to determine the efficacy of these FLT3 inhibitors and correlate it with patient outcomes. Results The different FLT3 compounds tested had a heterogenous effect on activated FLT3 levels. In some patients, ASP2215 caused a decrease in levels of activated FLT3 (indicated by brown nuclear staining), which corresponded to a decrease in leukemic blasts (Fig 1). Other patients, however, showed similar amounts of activated FLT3 both before and after treatment, which corresponded with no significant change in leukemic blasts. (Fig 2). Other FLT3 inhibitors also showed differences in their effects. Midostaurin reduced activated FLT3 levels, which correlated with a positive clinical response. In contrast, one patient receiving quizartinibshowed little to no decrease in activated FLT3 levels, despite remaining in clinical remission. This suggests that this FLT3 inhibitor may have alternative targets, such as the tyrosine kinases AXL or LTK. Conclusions The heterogeneity in the responses to ASP2215, midostaurin, and quizartinib suggests that there may be other targets for these compounds that are not currently accounted for in the clinical studies. Immunohistochemicalmeasurements of activated FLT3 in bone marrow sections before and after treatment with FLT3 inhibitors was not predictive for clinical response. Figure 1. Activated FLT3 levels in patient responsive to ASP2215; pre-treatment (A) and post-treatment (B) Figure 1. Activated FLT3 levels in patient responsive to ASP2215; pre-treatment (A) and post-treatment (B) Figure 2. Activated FLT3 levels in patient not responsive to ASP2215; pre-treatment (A) and post-treatment (B) Figure 2. Activated FLT3 levels in patient not responsive to ASP2215; pre-treatment (A) and post-treatment (B) Disclosures Larson: Pfizer: Consultancy; Ariad: Consultancy, Research Funding; Novartis: Consultancy, Research Funding; Bristol-Myers Squibb: Consultancy.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 34-35
Author(s):  
Jan Philipp Bewersdorf ◽  
Martin S. Tallman ◽  
Christina Cho ◽  
Amer M. Zeidan ◽  
Maximilian Stahl

Background: Prognosis of patients (pts) with acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS) who relapse after allogeneic hematopoietic cell transplant (allo-HCT) is extremely poor and strategies to reduce the risk of disease relapse are warranted. Hypomethylating agents (HMA) or FLT3 inhibitors have been used in several studies for relapse prevention following allo-HCT with mixed results leaving the question regarding the safety and efficacy of this strategy unanswered. Methods: We conducted a systematic review and meta-analysis and searched MEDLINE, EMBASE, Web of Science and CENTRAL from inception to 8/2020 for studies using the following combination of free-text terms linked by Boolean operators: [Acute myeloid leukemia OR AML OR MDS OR Myelodysplastic syndrome] AND [transplant OR allogeneic stem cell transplant OR hematopoietic stem cell transplantation] AND [maintenance OR maintenance therapy OR maintenance treatment]. Titles and abstracts were reviewed and excluded if they were review or basic research articles, not on post-allo-HCT maintenance in AML or MDS, no English full-text was available, or if they were clinical trials without published results. Full-texts were reviewed and excluded if (I) duplicate publications from the same patient cohort, (II) insufficient reporting of endpoints, (III) studies not on FLT3 inhibitors or HMA, (IV) case series with <5 pts, or (V) commentaries without original independent data. Studies using FLT3 inhibitors or HMA for pre-emptive treatment of imminent relapse based on positive minimal residual disease (MRD) testing were excluded. Outcomes of interest were the rates of 2-year overall survival (OS) and relapse-free survival (RFS) as well as incidence of acute and chronic graft-versus-host disease (GVHD). The study protocol was registered on PROSPERO (CRD42020187298). Results: The search strategy retrieved 1388 unique citations (Figure 1). After application of additional exclusion criteria, 22 studies were included in the meta-analysis. A total of 829 pts was included with 462 and 367 receiving post-allo-HCT treatment with FLT3 inhibitors or HMA, respectively. All pts treated with FLT3 inhibitors had AML, while 231 AML and 112 MDS pts were treated with HMA, respectively. Among studies on FLT3 inhibitors, sorafenib was used in 10 studies with midostaurin and quizartinib being used in 1 study each. Azacitidine was used in 8 studies, decitabine in 3 studies and 1 study used both azacitidine and decitabine. Patient, transplant, and treatment characteristics of the included studies are shown in Table 1. Reporting of outcomes was variable among the included studies (Figure 2;Panels A-D). Among pts treated with FLT3 inhibitors, 2-year OS and RFS rates were 81.7% (95% confidence interval [CI]: 70.0 - 89.5%), and 82.9% (95% CI: 76.9 - 87.5%), respectively. Acute and chronic GVHD occurred in 10.4% (95% CI: 0.5 - 73.3%) and 38.4% (95% CI: 13.4 - 71.5%) of pts, respectively. In HMA-treated pts, 2-year OS and RFS rates were 65.6% (95% CI: 54.7 - 75.1%) and 56.2% (42.4 - 69.2%), respectively. Acute and chronic GVHD occurred in 39.9% (95% CI: 29.2 - 51.6%) and 44.4% (95% CI: 34.1 - 55.2%) of pts, respectively. Study quality was limited by retrospective design employed by 10 studies, small sample sizes in some of the analyses, and heterogenous patient populations, reporting of outcomes, and transplant characteristics which were limiting cross-study comparisons. Discussion: Maintenance therapy with FLT3 inhibitors or HMA following allo-HCT in AML and MDS pts appears to be safe and can potentially be associated with prolonged RFS and OS although its efficacy needs to be verified in randomized trials. Careful patient selection is necessary as both the rates of GVHD and the burden on pts related to extended treatment following allo-HCT can be substantial. Selecting pts based on high-risk genetic (e.g. presence of TP53 mutations) and other disease characteristics (MRD-positivity at time of allo-HCT) could be options for patient selection but require additional validation. Various clinical trials investigating the role of post allo-HCT maintenance therapy are ongoing and our data could serve as a reference when interpreting the results of those trials. Disclosures Tallman: Daiichi-Sankyo: Membership on an entity's Board of Directors or advisory committees; Abbvie: Research Funding; Jazz Pharma: Membership on an entity's Board of Directors or advisory committees; Amgen: Research Funding; Orsenix: Research Funding; Cellerant: Research Funding; ADC Therapeutics: Research Funding; Bioline rx: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees; BioSight: Membership on an entity's Board of Directors or advisory committees, Research Funding; Rafael: Research Funding; Glycomimetics: Research Funding; Roche: Membership on an entity's Board of Directors or advisory committees; UpToDate: Patents & Royalties; Delta Fly Pharma: Membership on an entity's Board of Directors or advisory committees; Oncolyze: Membership on an entity's Board of Directors or advisory committees; Rigel: Membership on an entity's Board of Directors or advisory committees; KAHR: Membership on an entity's Board of Directors or advisory committees. Zeidan:Incyte: Consultancy, Honoraria, Research Funding; CCITLA: Other; Astex: Research Funding; Trovagene: Consultancy, Honoraria, Research Funding; Aprea: Research Funding; Seattle Genetics: Consultancy, Honoraria; ADC Therapeutics: Research Funding; Cardiff Oncology: Consultancy, Honoraria, Other; Ionis: Consultancy, Honoraria; Epizyme: Consultancy, Honoraria; Otsuka: Consultancy, Honoraria; Abbvie: Consultancy, Honoraria, Research Funding; Agios: Consultancy, Honoraria; Boehringer-Ingelheim: Consultancy, Honoraria, Research Funding; Novartis: Consultancy, Honoraria, Research Funding; Acceleron: Consultancy, Honoraria; Astellas: Consultancy, Honoraria; Daiichi Sankyo: Consultancy, Honoraria; Cardinal Health: Consultancy, Honoraria; Taiho: Consultancy, Honoraria; BeyondSpring: Consultancy, Honoraria; Leukemia and Lymphoma Society: Other; Takeda: Consultancy, Honoraria, Research Funding; Celgene / BMS: Consultancy, Honoraria, Research Funding; MedImmune/Astrazeneca: Research Funding; Jazz: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria, Research Funding.


Sign in / Sign up

Export Citation Format

Share Document